1
|
Calvi GDS, Cartaxo GNJ, Carretoni QL, da Silva ALM, de Moraes DN, Pradella JGDC, Costa MS. Inhibition of Development and Metabolism of Dual-Species Biofilms of Candida albicans and Candida krusei ( Pichia kudriavzevii) by Organoselenium Compounds. Pharmaceuticals (Basel) 2024; 17:1078. [PMID: 39204183 PMCID: PMC11359205 DOI: 10.3390/ph17081078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/07/2024] [Accepted: 08/14/2024] [Indexed: 09/03/2024] Open
Abstract
Although Candida albicans is the most frequently identified Candida species in clinical settings, a significant number of infections related to the non-albicans Candida (NAC) species, Candida krusei, has been reported. Both species are able to produce biofilms and have been an important resistance-related factor to antimicrobial resistance. In addition, the microbial relationship is common in the human body, contributing to the formation of polymicrobial biofilms. Considering the great number of reports showing the increase in cases of resistance to the available antifungal drugs, the development of new and effective antifungal agents is critical. The inhibitory effect of Organoselenium Compounds (OCs) on the development of Candida albicans and Candida krusei was recently demonstrated, supporting the potential of these compounds as efficient antifungal drugs. In addition, OCs were able to reduce the viability and the development of biofilms, a very important step in colonization and infection caused by fungi. Thus, the objective of this study was to investigate the effect of the Organoselenium Compounds (p-MeOPhSe)2, (PhSe)2, and (p-Cl-PhSe)2 on the development of dual-species biofilms of Candida albicans and Candida krusei produced using either RPMI-1640 or Sabouraud Dextrose Broth (SDB) media. The development of dual-species biofilms was evaluated by the determination of both metabolic activity, using a metabolic assay based on the reduction of XTT (2,3-bis(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide sodium salt) assay and identification of either Candida albicans and Candida krusei on CHROMagar Candida medium. Biofilm formation using RPMI-1640 was inhibited in 90, 55, and 20% by 30 µM (p-MeOPhSe)2, (PhSe)2, and (p-Cl-PhSe)2, respectively. However, biofilms produced using SDB presented an inhibition of 62, 30 and 15% in the presence of 30 µM (p-MeOPhSe)2, (PhSe)2, and (p-Cl-PhSe)2, respectively. The metabolic activity of 24 h biofilms was inhibited by 35, 30 and 20% by 30 µM (p-MeOPhSe)2, (PhSe)2, and (p-Cl-PhSe)2, respectively, with RPMI-1640; however, 24 h biofilms formed using SDB were not modified by the OCs. In addition, a great reduction in the number of CFUs of Candida albicans (93%) in biofilms produced using RPMI-1640 in the presence of 30 µM (p-MeOPhSe)2 was observed. However, biofilms formed using SDB and treated with 30 µM (p-MeOPhSe)2 presented a reduction of 97 and 69% in the number of CFUs of Candida albicans and Candida krusei, respectively. These results demonstrated that Organoselenium Compounds, mainly (p-MeOPhSe)2, are able to decrease the metabolic activity of dual-species biofilms by reducing both Candida albicans and Candida krusei cell number during biofilm formation using either RPMI-1640 or SDB. Taken together, these results demonstrated the potential of the OCs to inhibit the development of dual-species biofilms of Candida albicans and Candida krusei.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Maricilia Silva Costa
- Instituto de Pesquisa & Desenvolvimento—IP&D, Universidade do Vale do Paraíba—UNIVAP, Av. Shishima Hifumi, 2911, São José dos Campos 12244-390, SP, Brazil; (G.d.S.C.); (G.N.J.C.); (Q.L.C.); (A.L.M.d.S.); (D.N.d.M.); (J.G.d.C.P.)
| |
Collapse
|
2
|
Hassan AM, Khateb AM, Turkistani SA, Alhamdan MM, Garout RM, Dwivedi VD, Azhar EI. Structural analogs of 2-(4-fluorophenyl)-6-methyl-3-(pyridin-4-yl)pyrazolo[1,5-a]pyridine for targeting Candida albicans non-essential stress kinase Yck2 through protein-ligand binding and dynamics analysis. Front Chem 2024; 12:1430157. [PMID: 39193538 PMCID: PMC11347327 DOI: 10.3389/fchem.2024.1430157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/05/2024] [Indexed: 08/29/2024] Open
Abstract
The rise in drug-resistant fungal infections poses a significant public health concern, necessitating the development of new antifungal therapies. We aimed to address this challenge by targeting a yeast casein kinase of Candida albicans for antifungal drug development. The compound library contained 589 chemical structures similar to the previously identified kinase inhibitor GW461484A. Through virtual screening, four compounds with the PubChem IDs 102583821, 12982634, 102487860, and 86260205 were selected based on their binding energies. Hydrophobic bonds and van der Waals interactions stabilised the docked complexes. Comprehensive interaction studies and a 200-nanosecond molecular dynamics simulation suggested that these molecules can maintain stable interactions with the target, as evidenced by satisfactory RMSD and RMSF values. The Rg-RMSD-based Free Energy Landscape of these complexes indicated thermodynamic stability due to the presence of conformers with global minima. These promising findings highlight the potential for developing novel antifungal therapies targeting Yck2 in C. albicans. Further experimental validation is required to assess the efficacy of these compounds as antifungal agents. This research provides a significant step towards combating antifungal resistance and opens up a new avenue for drug discovery.
Collapse
Affiliation(s)
- Ahmed M. Hassan
- Special Infectious Agents Unit—BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Aiah M. Khateb
- Special Infectious Agents Unit—BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, Taibah University, Medina, Saudi Arabia
| | - Safaa A. Turkistani
- Special Infectious Agents Unit—BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Medical Laboratory Sciences Department, Fakeeh College for Medical Sciences, Jeddah, Saudi Arabia
| | - Meshari M. Alhamdan
- Special Infectious Agents Unit—BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Family Medicine Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Raed M. Garout
- Special Infectious Agents Unit—BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Molecular Diagnostics Laboratory, Clinical Laboratory Department, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Vivek Dhar Dwivedi
- Center for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Saveetha Medical College and Hospitals, Saveetha University, Chennai, India
- Bioinformatics Research Division, Quanta Calculus, Greater Noida, India
| | - Esam I. Azhar
- Special Infectious Agents Unit—BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Medical Laboratory Sciences Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
3
|
Walsh TJ. Meeting the Therapeutic Challenges of Emergent and Rare Invasive Fungal Diseases Through Novel Clinical Trial Designs. Open Forum Infect Dis 2024; 11:ofae257. [PMID: 38887484 PMCID: PMC11181194 DOI: 10.1093/ofid/ofae257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Indexed: 06/20/2024] Open
Abstract
Treatments for emerging and rare invasive fungal diseases (IFDs) represent a critical unmet medical need. For IFDs that occur less frequently than invasive aspergillosis, such as mucormycosis, hyalohyphomycosis, and phaeohyphomycosis, randomized controlled clinical trials are impractical and unlikely to meet urgent public health needs. Understanding regulatory approaches for approval of drugs for rare cancers and rare metabolic diseases could help meet the challenges of studying drugs for rare IFDs. A single-arm, controlled clinical trial with a high-quality external control(s), with confirmatory evidence from nonclinical studies, including pharmacokinetic/pharmacodynamic data in predictive animal models of the disease may support findings of effectiveness of new drugs and biologics. Control populations may include historical controls from published literature, patient registries, and/or contemporaneous external control groups. Continuous engagement among clinicians, industrial sponsors, and regulatory agencies to develop consensus on trial design and innovative development pathways for emergent and rare invasive fungal diseases is important.
Collapse
Affiliation(s)
- Thomas J Walsh
- Center for Innovative Therapeutics and Diagnostics, Office of the Director (citdx.org), Richmond, Virginia, USA
- Departments of Medicine and of Microbiology & Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
4
|
Puumala E, Sychantha D, Lach E, Reeves S, Nabeela S, Fogal M, Nigam A, Johnson JW, Aspuru-Guzik A, Shapiro RS, Uppuluri P, Kalyaanamoorthy S, Magolan J, Whitesell L, Robbins N, Wright GD, Cowen LE. Allosteric inhibition of tRNA synthetase Gln4 by N-pyrimidinyl-β-thiophenylacrylamides exerts highly selective antifungal activity. Cell Chem Biol 2024; 31:760-775.e17. [PMID: 38402621 PMCID: PMC11031294 DOI: 10.1016/j.chembiol.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/19/2023] [Accepted: 01/29/2024] [Indexed: 02/27/2024]
Abstract
Candida species are among the most prevalent causes of systemic fungal infections, which account for ∼1.5 million annual fatalities. Here, we build on a compound screen that identified the molecule N-pyrimidinyl-β-thiophenylacrylamide (NP-BTA), which strongly inhibits Candida albicans growth. NP-BTA was hypothesized to target C. albicans glutaminyl-tRNA synthetase, Gln4. Here, we confirmed through in vitro amino-acylation assays NP-BTA is a potent inhibitor of Gln4, and we defined how NP-BTA arrests Gln4's transferase activity using co-crystallography. This analysis also uncovered Met496 as a critical residue for the compound's species-selective target engagement and potency. Structure-activity relationship (SAR) studies demonstrated the NP-BTA scaffold is subject to oxidative and non-oxidative metabolism, making it unsuitable for systemic administration. In a mouse dermatomycosis model, however, topical application of the compound provided significant therapeutic benefit. This work expands the repertoire of antifungal protein synthesis target mechanisms and provides a path to develop Gln4 inhibitors.
Collapse
Affiliation(s)
- Emily Puumala
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - David Sychantha
- M.G. DeGroote Institute for Infectious Disease Research, David Braley Centre for Antibiotic Discovery, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Elizabeth Lach
- M.G. DeGroote Institute for Infectious Disease Research, David Braley Centre for Antibiotic Discovery, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Shawn Reeves
- Department of Chemistry, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Sunna Nabeela
- Division of Infectious Diseases, The Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles Medical Center, Torrance, CA 90502, USA
| | - Meea Fogal
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - AkshatKumar Nigam
- Department of Computer Science, Stanford University, Stanford, CA 94305, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Jarrod W Johnson
- M.G. DeGroote Institute for Infectious Disease Research, David Braley Centre for Antibiotic Discovery, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Alán Aspuru-Guzik
- Chemical Physics Theory Group, Department of Chemistry, University of Toronto Toronto, ON M5S 3H6, Canada; Department of Computer Science, University of Toronto, Toronto, ON M5S 2E4, Canada; Department of Chemical Engineering & Applied Chemistry, University of Toronto, Toronto, ON M5S 3E5, Canada; Department of Materials Science & Engineering, University of Toronto, Toronto, ON M5S 3E4, Canada; Vector Institute for Artificial Intelligence, Toronto, ON M5G 1M1, Canada; Lebovic Fellow, Canadian Institute for Advanced Research (CIFAR), Toronto, ON M5G 1M1, Canada; Acceleration Consortium, University of Toronto, Toronto, ON M5S 3H6, Canada
| | - Rebecca S Shapiro
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Priya Uppuluri
- Division of Infectious Diseases, The Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles Medical Center, Torrance, CA 90502, USA; David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | | | - Jakob Magolan
- M.G. DeGroote Institute for Infectious Disease Research, David Braley Centre for Antibiotic Discovery, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Luke Whitesell
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Gerard D Wright
- M.G. DeGroote Institute for Infectious Disease Research, David Braley Centre for Antibiotic Discovery, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
5
|
Hamid A, Mäser P, Mahmoud AB. Drug Repurposing in the Chemotherapy of Infectious Diseases. Molecules 2024; 29:635. [PMID: 38338378 PMCID: PMC10856722 DOI: 10.3390/molecules29030635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/18/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Repurposing is a universal mechanism for innovation, from the evolution of feathers to the invention of Velcro tape. Repurposing is particularly attractive for drug development, given that it costs more than a billion dollars and takes longer than ten years to make a new drug from scratch. The COVID-19 pandemic has triggered a large number of drug repurposing activities. At the same time, it has highlighted potential pitfalls, in particular when concessions are made to the target product profile. Here, we discuss the pros and cons of drug repurposing for infectious diseases and analyze different ways of repurposing. We distinguish between opportunistic and rational approaches, i.e., just saving time and money by screening compounds that are already approved versus repurposing based on a particular target that is common to different pathogens. The latter can be further distinguished into divergent and convergent: points of attack that are divergent share common ancestry (e.g., prokaryotic targets in the apicoplast of malaria parasites), whereas those that are convergent arise from a shared lifestyle (e.g., the susceptibility of bacteria, parasites, and tumor cells to antifolates due to their high rate of DNA synthesis). We illustrate how such different scenarios can be capitalized on by using examples of drugs that have been repurposed to, from, or within the field of anti-infective chemotherapy.
Collapse
Affiliation(s)
- Amal Hamid
- Faculty of Pharmacy, University of Khartoum, Khartoum 11111, Sudan;
| | - Pascal Mäser
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Allschwil, 4123 Basel, Switzerland
- Faculty of Science, University of Basel, 4001 Basel, Switzerland
| | - Abdelhalim Babiker Mahmoud
- Faculty of Pharmacy, University of Khartoum, Khartoum 11111, Sudan;
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland, 66123 Saarbruecken, Germany
- Department of Microbial Drugs, Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany
| |
Collapse
|
6
|
Liu JC, Yang J, Lei SX, Wang MF, Ma YN, Yang R. Natural phytoalexins inspired the discovery of new biphenyls as potent antifungal agents for treatment of invasive fungal infections. Eur J Med Chem 2023; 261:115842. [PMID: 37788549 DOI: 10.1016/j.ejmech.2023.115842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/11/2023] [Accepted: 09/26/2023] [Indexed: 10/05/2023]
Abstract
With the aim of discovering novel and effective antifungal agents derived from natural sources, a series of new biphenyls based on natural biphenyl phytoalexins were designed, synthesized and evaluated for their antifungal activities against four invasive fungi. By modifying the two benzene rings of noraucuparin, a well-known biphenyl phytoantitoxin, some promising compounds with remarkable antifungal activity were discovered. Notably, compounds 23a, 23e and 23h exhibited potent activities and a broad antifungal spectrum with low MICs of 0.25-16 μg/mL, which were 8-256-fold more potent than that of the lead compound noraucuparin. Particularly, they displayed comparable potency to the positive control amphotericin B against Cryptococcus neoformans. Some interesting structure-activity relationships have also been discussed. Preliminary mechanism studies revealed that compound 23h might achieve its rapid fungicidal activity by disrupting the fungal cell membrane. Moreover, compound 23h exhibited significant inhibition against some virulence factors of Cryptococcus neoformans, low toxicity to normal human cells, as well as favorable pharmacokinetic and drug-like properties. The above results evidenced that the development of new antifungal candidates derived from natural phytoalexins was a bright and promising strategy.
Collapse
Affiliation(s)
- Jian-Chuan Liu
- College of Materials, Chemistry & Chemical Engineering, Chengdu University of Technology, Chengdu, 610059, China
| | - Jian Yang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Shu-Xin Lei
- College of Ecological Environment, Chengdu University of Technology, Chengdu, 610059, China
| | - Ming-Fan Wang
- College of Materials, Chemistry & Chemical Engineering, Chengdu University of Technology, Chengdu, 610059, China
| | - Yan-Ni Ma
- Henan Academy of Sciences, Zhengzhou, 450002, China; Medical School, Huanghe Science & Technology University, Zhengzhou, 450063, China
| | - Rui Yang
- College of Materials, Chemistry & Chemical Engineering, Chengdu University of Technology, Chengdu, 610059, China.
| |
Collapse
|
7
|
Ajetunmobi OH, Badali H, Romo JA, Ramage G, Lopez-Ribot JL. Antifungal therapy of Candida biofilms: Past, present and future. Biofilm 2023; 5:100126. [PMID: 37193227 PMCID: PMC10182175 DOI: 10.1016/j.bioflm.2023.100126] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 05/18/2023] Open
Abstract
Virtually all Candida species linked to clinical candidiasis are capable of forming highly resistant biofilms on different types of surfaces, which poses an additional significant threat and further complicates therapy of these infections. There is a scarcity of antifungal agents, and their effectiveness, particularly against biofilms, is limited. Here we provide a historical perspective on antifungal agents and therapy of Candida biofilms. As we reflect upon the past, consider the present, and look towards the future of antifungal therapy of Candida biofilms, we believe that there are reasons to remain optimistic, and that the major challenges of Candida biofilm therapy can be conquered within a reasonable timeframe.
Collapse
Affiliation(s)
- Olabayo H. Ajetunmobi
- Department of Molecular Microbiology & Immunology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Hamid Badali
- Department of Molecular Microbiology & Immunology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Jesus A. Romo
- Department of Molecular Microbiology & Immunology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Gordon Ramage
- Glasgow Biofilm Research Network, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Jose L. Lopez-Ribot
- Department of Molecular Microbiology & Immunology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, USA
- Corresponding author. Department of Molecular Microbiology & Immunology, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA.
| |
Collapse
|
8
|
Spadari CDC, Borba-Santos LP, Rozental S, Ishida K. Miltefosine repositioning: A review of potential alternative antifungal therapy. J Mycol Med 2023; 33:101436. [PMID: 37774486 DOI: 10.1016/j.mycmed.2023.101436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 09/17/2023] [Accepted: 09/19/2023] [Indexed: 10/01/2023]
Abstract
Fungal infections are a global health problem with high mortality and morbidity rates. Available antifungal agents have high toxicity and pharmacodynamic and pharmacokinetic limitations. Moreover, the increased incidence of antifungal-resistant isolates and the emergence of intrinsically resistant species raise concerns about seeking alternatives for efficient antifungal therapy. In this context, we review literature data addressing the potential action of miltefosine (MFS), an anti-Leishmania and anticancer agent, as a repositioning drug for antifungal treatment. Here, we highlight the in vitro and in vivo data, MFS possible mechanisms of action, case reports, and nanocarrier-mediated MFS delivery, focusing on fungal infection therapy. Finally, many studies have demonstrated the promising antifungal action of MFS in vitro, but there is little or no data on antifungal activity in vertebrate animal models and clinical trials, so have a need to develop more research for the repositioning of MFS as an antifungal therapy.
Collapse
Affiliation(s)
| | - Luana Pereira Borba-Santos
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sonia Rozental
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Kelly Ishida
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
9
|
Ramage G, Borghi E, Rodrigues CF, Kean R, Williams C, Lopez-Ribot J. Our current clinical understanding of Candida biofilms: where are we two decades on? APMIS 2023; 131:636-653. [PMID: 36932821 DOI: 10.1111/apm.13310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 03/12/2023] [Indexed: 03/19/2023]
Abstract
Clinically we have been aware of the concept of Candida biofilms for many decades, though perhaps without the formal designation. Just over 20 years ago the subject emerged on the back of progress made from the bacterial biofilms, and academic progress pace has continued to mirror the bacterial biofilm community, albeit at a decreased volume. It is apparent that Candida species have a considerable capacity to colonize surfaces and interfaces and form tenacious biofilm structures, either alone or in mixed species communities. From the oral cavity, to the respiratory and genitourinary tracts, wounds, or in and around a plethora of biomedical devices, the scope of these infections is vast. These are highly tolerant to antifungal therapies that has a measurable impact on clinical management. This review aims to provide a comprehensive overight of our current clinical understanding of where these biofilms cause infections, and we discuss existing and emerging antifungal therapies and strategies.
Collapse
Affiliation(s)
- Gordon Ramage
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, UK
- Study Group for Biofilms (ESGB), European Society for Clinical Microbiology and Infectious Disease, Basel, Switzerland
| | - Elisa Borghi
- Study Group for Biofilms (ESGB), European Society for Clinical Microbiology and Infectious Disease, Basel, Switzerland
- Department of Health Sciences, San Paolo Medical School, Università Degli Studi di Milano, Milan, Italy
| | - Célia Fortuna Rodrigues
- Study Group for Biofilms (ESGB), European Society for Clinical Microbiology and Infectious Disease, Basel, Switzerland
- LEPABE-Department of Chemical Engineering, Faculty of Engineering, Cooperativa de Ensino Superior Politécnico e Universitário-CESPU, Gandra, Portugal
- ALiCE-Associate Laboratory in Chemical Engineering, Faculty of Engineering, Cooperativa de Ensino Superior Politécnico e Universitário-CESPU, Gandra, Portugal
- TOXRUN-Toxicology Research Unit, Cooperativa de Ensino Superior Politécnico e Universitário-CESPU, Gandra, Portugal
| | - Ryan Kean
- Study Group for Biofilms (ESGB), European Society for Clinical Microbiology and Infectious Disease, Basel, Switzerland
- Department of Biological Sciences, Glasgow Caledonian University, Glasgow, UK
| | - Craig Williams
- Study Group for Biofilms (ESGB), European Society for Clinical Microbiology and Infectious Disease, Basel, Switzerland
- Microbiology Department, Morecambe Bay NHS Trust, Lancaster, UK
| | - Jose Lopez-Ribot
- Department of Biology and the South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
10
|
Ajetunmobi OH, Chaturvedi AK, Badali H, Vaccaro A, Najvar L, Wormley FL, Wiederhold NP, Patterson TF, Lopez-Ribot JL. Screening the medicine for malaria venture's Pandemic Response Box to identify novel inhibitors of Candida albicans and Candida auris biofilm formation. APMIS 2023; 131:613-625. [PMID: 37337909 PMCID: PMC10592529 DOI: 10.1111/apm.13342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/21/2023]
Abstract
Candida spp. are opportunistic yeasts capable of forming biofilms, which contribute to resistance, increasing the urgency for new effective antifungal therapies. Repurposing existing drugs could significantly accelerate the development of novel therapies against candidiasis. We screened the Pandemic Response Box containing 400 diverse drug-like molecules active against bacteria, viruses or fungi, for inhibitors of Candida albicans and Candida auris biofilm formation. Initial hits were identified based on the demonstration of >70% inhibitory activity. Dose-response assays were used to confirm the antifungal activity of initial hits and establish their potency. The spectrum of antifungal activity of the leading compounds was determined against a panel of medically important fungi, and the in vivo activity of the leading repositionable agent was evaluated in murine models of C. albicans and C. auris systemic candidiasis. The primary screening identified 20 hit compounds, and their antifungal activity and potency against C. albicans and C. auris were validated using dose-response measurements. From these experiments, the rapalog everolimus, emerged as the leading repositionable candidate. Everolimus displayed potent antifungal activity against different Candida spp., but more moderate levels of activity against filamentous fungi. Treatment with everolimus increased survival of mice infected with C. albicans, but not those with C. auris. The screening of the Pandemic Response Box resulted in the identification of several drugs with novel antifungal activity, with everolimus emerging as the main repositionable candidate. Further in vitro and in vivo studies are needed to confirm its potential therapeutic use.
Collapse
Affiliation(s)
- Olabayo H. Ajetunmobi
- Department of Molecular Microbiology & Immunology, and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, Texas, USA
| | - Ashok K. Chaturvedi
- Department of Molecular Microbiology & Immunology, and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, Texas, USA
| | - Hamid Badali
- Department of Molecular Microbiology & Immunology, and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, Texas, USA
| | - Alessandra Vaccaro
- Department of Molecular Microbiology & Immunology, and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, Texas, USA
| | - Laura Najvar
- Department of Medicine, Division of Infectious Diseases, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Floyd L. Wormley
- Department of Biology, Texas Christian University, Fort Worth, Texas, USA
| | - Nathan P. Wiederhold
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Thomas F. Patterson
- Department of Medicine, Division of Infectious Diseases, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Jose L. Lopez-Ribot
- Department of Molecular Microbiology & Immunology, and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
11
|
Ajetunmobi OH, Wall G, Vidal Bonifacio B, Martinez Delgado LA, Chaturvedi AK, Najvar LK, Wormley FL, Patterson HP, Wiederhold NP, Patterson TF, Lopez-Ribot JL. High-Throughput Screening of the Repurposing Hub Library to Identify Drugs with Novel Inhibitory Activity against Candida albicans and Candida auris Biofilms. J Fungi (Basel) 2023; 9:879. [PMID: 37754987 PMCID: PMC10532723 DOI: 10.3390/jof9090879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/18/2023] [Accepted: 08/25/2023] [Indexed: 09/28/2023] Open
Abstract
Candidiasis is one of the most frequent nosocomial infections affecting an increasing number of at-risk patients. Candida albicans remains the most frequent causative agent of candidiasis, but, in the last decade, C. auris has emerged as a formidable multi-drug-resistant pathogen. Both species are fully capable of forming biofilms, which contribute to resistance, increasing the urgency for new effective antifungal therapies. Repurposing existing drugs could significantly accelerate the development of novel therapies against candidiasis. Here, we have screened the Repurposing Hub library from the Broad Institute, containing over 6000 compounds, in search for inhibitors of C. albicans and C. auris biofilm formation. The primary screen identified 57 initial hits against C. albicans and 33 against C. auris. Confirmatory concentration-dependent assays were used to validate the activity of the initial hits and, at the same time, establish their anti-biofilm potency. Based on these results, ebselen, temsirolimus, and compound BAY 11-7082 emerged as the leading repositionable compounds. Subsequent experiments established their spectrum of antifungal activity against yeasts and filamentous fungi. In addition, their in vivo activity was examined in the murine models of hematogenously disseminated C. albicans and C. auris infections. Although promising, further in vitro and in vivo studies are needed to confirm their potential use for the therapy of candidiasis and possibly other fungal infections.
Collapse
Affiliation(s)
- Olabayo H. Ajetunmobi
- Department of Molecular Microbiology & Immunology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, USA; (O.H.A.); (A.K.C.)
| | - Gina Wall
- Department of Molecular Microbiology & Immunology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, USA; (O.H.A.); (A.K.C.)
| | - Bruna Vidal Bonifacio
- Department of Molecular Microbiology & Immunology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, USA; (O.H.A.); (A.K.C.)
| | | | - Ashok K. Chaturvedi
- Department of Molecular Microbiology & Immunology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, USA; (O.H.A.); (A.K.C.)
| | - Laura K. Najvar
- Department of Medicine, Division of Infectious Diseases, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (L.K.N.); (T.F.P.)
| | - Floyd L. Wormley
- Department of Biology, Texas Christian University, Fort Worth, TX 76129, USA;
| | - Hoja P. Patterson
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (H.P.P.); (N.P.W.)
| | - Nathan P. Wiederhold
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (H.P.P.); (N.P.W.)
| | - Thomas F. Patterson
- Department of Medicine, Division of Infectious Diseases, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (L.K.N.); (T.F.P.)
| | - Jose L. Lopez-Ribot
- Department of Molecular Microbiology & Immunology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, USA; (O.H.A.); (A.K.C.)
| |
Collapse
|
12
|
Zhu P, Li Y, Guo T, Liu S, Tancer RJ, Hu C, Zhao C, Xue C, Liao G. New antifungal strategies: drug combination and co-delivery. Adv Drug Deliv Rev 2023; 198:114874. [PMID: 37211279 DOI: 10.1016/j.addr.2023.114874] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/12/2023] [Accepted: 05/14/2023] [Indexed: 05/23/2023]
Abstract
The growing occurrence of invasive fungal infections and the mounting rates of drug resistance constitute a significant menace to human health. Antifungal drug combinations have garnered substantial interest for their potential to improve therapeutic efficacy, reduce drug doses, reverse, or ameliorate drug resistance. A thorough understanding of the molecular mechanisms underlying antifungal drug resistance and drug combination is key to developing new drug combinations. Here we discuss the mechanisms of antifungal drug resistance and elucidate how to discover potent drug combinations to surmount resistance. We also examine the challenges encountered in developing such combinations and discuss prospects, including advanced drug delivery strategies.
Collapse
Affiliation(s)
- Ping Zhu
- State Key Laboratory of Silkworm Genome Biology, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400700, China
| | - Yan Li
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Ting Guo
- State Key Laboratory of Silkworm Genome Biology, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400700, China
| | - Simei Liu
- Department of Traditional Chinese Medicine, Chongqing College of Traditional Chinese Medicine, Chongqing 402760, China; Institute of Pharmacology and Toxicology, Chongqing Academy of Chinese Materia Medica, Chongqing 400065, China
| | - Robert J Tancer
- Public Health Research Institute and Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Changhua Hu
- State Key Laboratory of Silkworm Genome Biology, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400700, China
| | - Chengzhi Zhao
- Chongqing Health Center for Women and Children, Chongqing, 400700, PR China.
| | - Chaoyang Xue
- Public Health Research Institute and Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Guojian Liao
- State Key Laboratory of Silkworm Genome Biology, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400700, China.
| |
Collapse
|
13
|
Bai YB, Zhang M, Li D, Zhao Y, Huang LZ, Gao JM. Synthesis and Antifungal Activity of Derivatives of the Natural Product Griseofulvin against Phytopathogenic Fungi. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:6236-6248. [PMID: 37061927 DOI: 10.1021/acs.jafc.2c09037] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Natural products are important sources for the discovery of new pesticides. Chemical synthesis and structural modification can lead to pesticides. Despite abundant research in fungicide discovery for crop protection, there is an emerging need for the development of novel antifungal agrochemicals. Herein, 39 diversified griseofulvin derivatives were effectively synthesized from the natural product griseofulvin by diversity-oriented synthesis through the reactions of demethylation, ammonolysis, methylation, nitration, acylation, reduction, and chlorination. Among them, 31 derivatives were novel. All structures were characterized by 1H NMR, 13C NMR, and high-resolution mass spectrometry (HR-MS), and the antifungal activity was investigated against five phytopathogenic fungi. Compounds 5h and 5l had excellent activity against Botrytis cinerea (5h, IC50 = 17.29 ± 0.64 μg/mL) and Alternaria solani (5l, IC50 = 22.52 ± 0.79 μg/mL), respectively. Compound 9 exhibited the more promising activities against three target fungi, especially against Colletotrichum gloeosporioides (IC50 = 7.24 ± 0.66 μg/mL), which is obviously better than positive control hymexazol, thifluzamide, and parent compound griseofulvin. In addition, compound 10 showed significant and extensive activities against four target fungi Cytospora sp. (IC50 = 18.72 ± 0.35 μg/mL), C. gloeosporioides (IC50 = 31.39 ± 1.48 μg/mL), A. solani (IC50 = 40.82 ± 1.04 μg/mL), and Fusarium solani (IC50 = 36.81 ± 0.82 μg/mL). Unexpectedly, 11 and 12, the chlorinated products of compound 9, exhibited the most promising activity against C. gloeosporioides (IC50 = 4.48 ± 0.54 μg/mL for 11, 2.24 ± 0.76 μg/mL for 12). Furthermore, 12 showed remarkable activity against Cytospora sp. (IC50 = 5.85 ± 0.72 μg/mL). Additionally, in vivo antifungal activity against C. gloeosporioides, homology modeling, and docking analysis of 11, 12, and griseofulvin were conducted. All results indicated that 11 and 12 had potency as antifungal agents against C. gloeosporioides, and the modifications of the 2' and 4' positions of griseofulvin should be further explored for higher-activity lead compounds or potential agricultural fungicides.
Collapse
Affiliation(s)
- Yu-Bin Bai
- Shaanxi Key Laboratory of Chemical Reaction Engineering, College of Chemistry and Chemical Engineering, Yan'an University, Yan'an, Shaanxi 716000, People's Republic of China
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, People's Republic of China
| | - Meng Zhang
- Shaanxi Key Laboratory of Chemical Reaction Engineering, College of Chemistry and Chemical Engineering, Yan'an University, Yan'an, Shaanxi 716000, People's Republic of China
| | - Ding Li
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, People's Republic of China
| | - Yu Zhao
- Shaanxi Key Laboratory of Chemical Reaction Engineering, College of Chemistry and Chemical Engineering, Yan'an University, Yan'an, Shaanxi 716000, People's Republic of China
| | - Liang-Zhu Huang
- Shaanxi Key Laboratory of Chemical Reaction Engineering, College of Chemistry and Chemical Engineering, Yan'an University, Yan'an, Shaanxi 716000, People's Republic of China
| | - Jin-Ming Gao
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, People's Republic of China
| |
Collapse
|
14
|
de Oliveira H, Bezerra BT, Rodrigues ML. Antifungal Development and the Urgency of Minimizing the Impact of Fungal Diseases on Public Health. ACS BIO & MED CHEM AU 2023; 3:137-146. [PMID: 37101810 PMCID: PMC10125384 DOI: 10.1021/acsbiomedchemau.2c00055] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 04/28/2023]
Abstract
Fungal infections are a major public health problem resulting from the lack of public policies addressing these diseases, toxic and/or expensive therapeutic tools, scarce diagnostic tests, and unavailable vaccines. In this Perspective, we discuss the need for novel antifungal alternatives, highlighting new initiatives based on drug repurposing and the development of novel antifungals.
Collapse
Affiliation(s)
| | - Bárbara T. Bezerra
- Instituto
Carlos Chagas, Fundação Oswaldo Cruz (Fiocruz), Curitiba81310-020, Brazil
| | - Marcio L. Rodrigues
- Instituto
Carlos Chagas, Fundação Oswaldo Cruz (Fiocruz), Curitiba81310-020, Brazil
- Instituto
de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro, Rio de Janeiro21941-902, Brazil
| |
Collapse
|
15
|
Ghobadi E, Hashemi SM, Fakhim H, Hosseini-Khah Z, Badali H, Emami S. Design, synthesis and biological activity of hybrid antifungals derived from fluconazole and mebendazole. Eur J Med Chem 2023; 249:115146. [PMID: 36709648 DOI: 10.1016/j.ejmech.2023.115146] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 01/22/2023]
Abstract
A novel series of triazole alcohol antifungals bearing a 5-benzoylbenzimidazol-2-ylthio side chain have been designed and synthesized as hybrids of fluconazole (a typical triazole antifungal) and mebendazole (an anthelmintic agent with antifungal activity). The title compounds were synthesized via the reaction of an appropriate oxirane and desired 2-mercaptobenzimidazole. Although there was possibility for formation of different N-substituted or S-substituted products, the structures of final compounds were assigned as thioether congeners by using 13C NMR spectroscopy. The SAR analysis of the primary lead compounds (series A) was conducted by simplifying the 5-benzoylbenzimidazol-2-ylthio residue to the benzimidazol-2-ylthio (series B) or benzothiazol-2-ylthio side chain (series C), and modification of halogen substituent on the phenethyl-triazole scaffold. In general, series A (compounds 4a-e) containing 5-benzoylbenzimidazole scaffold showed better antifungal activity against Candida spp. and Cryptococcus neoformans than related benzimidazole and benzothiazole derivatives. The better results were obtained with the 4-chloro derivative 4b displaying MICs <0.063-1 μg/mL. Although, removing benzoyl group from compound 4b had negative effect on the activity, optimization of phenethyl-triazole scaffold by desired halogen substituent resulted in compound 5c being as potent as 4b. In vitro and in silico ADMET evaluations of the most promising compounds 4b and 5c indicated that the selected compounds have desirable ADMET properties in comparison to standard drug fluconazole. Docking simulation study demonstrated that the benzimidazol-2-ylthio moiety is responsible for the potent antifungal activity of these compounds.
Collapse
Affiliation(s)
- Elham Ghobadi
- Department of Medicinal Chemistry and Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran; Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyedeh Mahdieh Hashemi
- Department of Medicinal Chemistry and Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hamed Fakhim
- Infectious Diseases and Tropical Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Hosseini-Khah
- Diabetes Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hamid Badali
- Invasive Fungi Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran; Department of Molecular Microbiology & Immunology, and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Saeed Emami
- Department of Medicinal Chemistry and Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
16
|
Aydin M, Ozturk A, Duran T, Ozmen UO, Sumlu E, Ayan EB, Korucu EN. In vitro antifungal and antibiofilm activities of novel sulfonyl hydrazone derivatives against Candida spp. J Mycol Med 2023; 33:101327. [PMID: 36272382 DOI: 10.1016/j.mycmed.2022.101327] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/26/2022] [Accepted: 08/15/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND The aim of this study was to investigate the antifungal and antibiofilm activity of the new sulfonyl hydrazones compound derived from sulphonamides. METHODS In this study, new sulfonyl hydrazone series were synthesized via a green chemistry method. The structures of the synthesized compounds were characterized by elemental analyses and spectroscopic methods. The antifungal activities of the Anaf compounds against Candida strains under planktonic conditions were tested. The biofilm-forming ability of Candida strains was determined and the inhibitory effects of Anaf compounds on Candida biofilms compared with fluconazole were measured by MTT assay. Expression analysis of biofilm-related genes was investigated with qRT-PCR. The statistical analysis was performed using a one-way ANOVA test. CANDIDA: strains was determined and the inhibitory effects of Anaf compounds on Candida biofilms compared with fluconazole were measured by MTT assay. Expression analysis of biofilm-related genes was investigated with qRT-PCR. The statistical analysis was performed using a one-way ANOVA test. RESULTS A total of 16 (45.7%) out of 35 Candida isolates were determined as strong biofilm producers in this study. C. albicans was the most biofilm producer, followed by C. krusei and C. lusitaniae. The Anaf compounds had a broad spectrum of activity with MIC values ranging from 4 μg/ml to 64 μg/ml. Our data indicated that the Anaf compound had a significant effect on inhibiting biofilm formation in both fluconazole-susceptible and -resistant strains. The expression levels of hypha-specific genes als3, hwp1, ece1 and sap5 were downregulated by Anaf compounds. CONCLUSIONS Our study revealed that the Anaf compounds had antifungal activity and inhibited fungal biofilms, which may be related to the suppression of C. albicans adherence and hyphal formation. These results suggest that Anaf compounds may have therapeutic potential for the treatment and prevention of biofilm-associated Candida infections.
Collapse
Affiliation(s)
- Merve Aydin
- Department of Medical Microbiology, Faculty of Medicine, KTO Karatay University, Konya, Turkey.
| | - Ali Ozturk
- Department of Medical Microbiology, Faculty of Medicine, Niğde Ömer Halisdemir University, Niğde, Turkey
| | - Tugce Duran
- Department of Medical Genetics, Faculty of Medicine, KTO Karatay University, Konya, Turkey
| | | | - Esra Sumlu
- Department of Medical Pharmacology, Faculty of Medicine, KTO Karatay University, Konya, Turkey
| | - Esra Bilen Ayan
- Department of Chemistry, Faculty of Science, Gazi University, Ankara, Turkey
| | - Emine Nedime Korucu
- Department of Molecular Biology and Genetics, Faculty of Science, Necmettin Erbakan University, Konya, Turkey
| |
Collapse
|
17
|
Bedoya-Cardona JE, Rubio-Carrasquilla M, Ramírez-Velásquez IM, Valdés-Tresanco MS, Moreno E. Identifying Potential Molecular Targets in Fungi Based on (Dis)Similarities in Binding Site Architecture with Proteins of the Human Pharmacolome. Molecules 2023; 28:molecules28020692. [PMID: 36677748 PMCID: PMC9860719 DOI: 10.3390/molecules28020692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/23/2022] [Accepted: 01/04/2023] [Indexed: 01/13/2023] Open
Abstract
Invasive fungal infections represent a public health problem that worsens over the years with the increasing resistance to current antimycotic agents. Therefore, there is a compelling medical need of widening the antifungal drug repertoire, following different methods such as drug repositioning, identification and validation of new molecular targets and developing new inhibitors against these targets. In this work we developed a structure-based strategy for drug repositioning and new drug design, which can be applied to infectious fungi and other pathogens. Instead of applying the commonly accepted off-target criterion to discard fungal proteins with close homologues in humans, the core of our approach consists in identifying fungal proteins with active sites that are structurally similar, but preferably not identical to binding sites of proteins from the so-called "human pharmacolome". Using structural information from thousands of human protein target-inhibitor complexes, we identified dozens of proteins in fungal species of the genera Histoplasma, Candida, Cryptococcus, Aspergillus and Fusarium, which might be exploited for drug repositioning and, more importantly, also for the design of new fungus-specific inhibitors. As a case study, we present the in vitro experiments performed with a set of selected inhibitors of the human mitogen-activated protein kinases 1/2 (MEK1/2), several of which showed a marked cytotoxic activity in different fungal species.
Collapse
Affiliation(s)
| | - Marcela Rubio-Carrasquilla
- Facultad de Ciencias Básicas, Universidad de Medellín, Medellin 050026, Colombia
- Corporación para Investigaciones Biológicas, Medellin 050034, Colombia
| | - Iliana M. Ramírez-Velásquez
- Facultad de Ciencias Básicas, Universidad de Medellín, Medellin 050026, Colombia
- Instituto Tecnológico Metropolitano, Medellin 050034, Colombia
| | | | - Ernesto Moreno
- Facultad de Ciencias Básicas, Universidad de Medellín, Medellin 050026, Colombia
- Correspondence:
| |
Collapse
|
18
|
Gorris ME, Ardon-Dryer K, Campuzano A, Castañón-Olivares LR, Gill TE, Greene A, Hung CY, Kaufeld KA, Lacy M, Sánchez-Paredes E. Advocating for Coccidioidomycosis to Be a Reportable Disease Nationwide in the United States and Encouraging Disease Surveillance across North and South America. J Fungi (Basel) 2023; 9:83. [PMID: 36675904 PMCID: PMC9863933 DOI: 10.3390/jof9010083] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/20/2022] [Accepted: 12/20/2022] [Indexed: 01/07/2023] Open
Abstract
Coccidioidomycosis (Valley fever) has been a known health threat in the United States (US) since the 1930s, though not all states are currently required to report disease cases. Texas, one of the non-reporting states, is an example of where both historical and contemporary scientific evidence define the region as endemic, but we don't know disease incidence in the state. Mandating coccidioidomycosis as a reportable disease across more US states would increase disease awareness, improve clinical outcomes, and help antifungal drug and vaccine development. It would also increase our understanding of where the disease is endemic and the relationships between environmental conditions and disease cases. This is true for other nations in North and South America that are also likely endemic for coccidioidomycosis, especially Mexico. This commentary advocates for US state and territory epidemiologists to define coccidioidomycosis as a reportable disease and encourages disease surveillance in other endemic regions across North and South America in order to protect human health and reduce disease burden.
Collapse
Affiliation(s)
- Morgan E. Gorris
- Information Systems and Modeling, Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | - Karin Ardon-Dryer
- Department of Geosciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Althea Campuzano
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Laura R. Castañón-Olivares
- Unidad de Micología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Thomas E. Gill
- Environmental Science and Engineering Program, University of Texas at El Paso, El Paso, TX 79968, USA
- Department of Earth, Environmental and Resource Sciences, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Andrew Greene
- Statistical Sciences, Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | - Chiung-Yu Hung
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Kimberly A. Kaufeld
- Statistical Sciences, Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | - Mark Lacy
- Infectious Disease, Pediatrics, Internal Medicine, University of New Mexico Health, Albuquerque, NM 87106, USA
| | - Edith Sánchez-Paredes
- Unidad de Micología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
19
|
Ajetunmobi OH, Wall G, Bonifacio BV, Montelongo-Jauregui D, Lopez-Ribot JL. A 384-Well Microtiter Plate Model for Candida Biofilm Formation and Its Application to High-Throughput Screening. Methods Mol Biol 2023; 2658:53-64. [PMID: 37024695 DOI: 10.1007/978-1-0716-3155-3_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
Candidiasis, infections caused by Candida spp., represents one of the most common nosocomial infections afflicting an expanding number of compromised patients. Antifungal therapeutic options are few and show limited efficacy. Moreover, biofilm formation is frequently associated with different manifestations of candidiasis and further complicates therapy. Thus, there is an urgent need for new effective therapeutic agents, particularly those with anti-biofilm activity. Here we describe the development of a novel, simple, fast, economical, and highly reproducible 384-well microtiter plate model for the formation of both Candida albicans and Candida auris biofilms and its application in high-throughput screening (HTS) techniques.
Collapse
Affiliation(s)
- Olabayo H Ajetunmobi
- Department of Molecular Microbiology and Immunology, and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Gina Wall
- Department of Molecular Microbiology and Immunology, and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Bruna V Bonifacio
- Department of Molecular Microbiology and Immunology, and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Daniel Montelongo-Jauregui
- Department of Molecular Microbiology and Immunology, and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Jose L Lopez-Ribot
- Department of Molecular Microbiology and Immunology, and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
20
|
Drug repurposing strategy II: from approved drugs to agri-fungicide leads. J Antibiot (Tokyo) 2023; 76:131-182. [PMID: 36707717 PMCID: PMC9880955 DOI: 10.1038/s41429-023-00594-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/22/2022] [Accepted: 12/27/2022] [Indexed: 01/28/2023]
Abstract
Epidemic diseases of crops caused by fungi deeply affected the course of human history and processed a major restriction on social and economic development. However, with the enormous misuse of existing antimicrobial drugs, an increasing number of fungi have developed serious resistance to them, making the diseases caused by pathogenic fungi even more challenging to control. Drug repurposing is an attractive alternative, it requires less time and investment in the drug development process than traditional R&D strategies. In this work, we screened 600 existing commercially available drugs, some of which had previously unknown activity against pathogenic fungi. From the primary screen at a fixed concentration of 100 μg/mL, 120, 162, 167, 85, 102, and 82 drugs were found to be effective against Rhizoctonia solani, Sclerotinia sclerotiorum, Botrytis cinerea, Phytophthora capsici, Fusarium graminearum and Fusarium oxysporum, respectively. They were divided into nine groups lead compounds, including quinoline alkaloids, benzimidazoles/carbamate esters, azoles, isothiazoles, pyrimidines, pyridines, piperidines/piperazines, ionic liquids and miscellaneous group, and simple structure-activity relationship analysis was carried out. Comparison with fungicides to identify the most promising drugs or lead structures for the development of new antifungal agents in agriculture.
Collapse
|
21
|
Vu K, Buckley BJ, Bujaroski RS, Blumwald E, Kelso MJ, Gelli A. Antifungal activity of 6-substituted amiloride and hexamethylene amiloride (HMA) analogs. Front Cell Infect Microbiol 2023; 13:1101568. [PMID: 36923593 PMCID: PMC10009331 DOI: 10.3389/fcimb.2023.1101568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/20/2023] [Indexed: 02/19/2023] Open
Abstract
Fungal infections have become an increasing threat as a result of growing numbers of susceptible hosts and diminishing effectiveness of antifungal drugs due to multi-drug resistance. This reality underscores the need to develop novel drugs with unique mechanisms of action. We recently identified 5-(N,N-hexamethylene)amiloride (HMA), an inhibitor of human Na+/H+ exchanger isoform 1, as a promising scaffold for antifungal drug development. In this work, we carried out susceptibility testing of 45 6-substituted HMA and amiloride analogs against a panel of pathogenic fungi. A series of 6-(2-benzofuran)amiloride and HMA analogs that showed up to a 16-fold increase in activity against Cryptococcus neoformans were identified. Hits from these series showed broad-spectrum activity against both basidiomycete and ascomycete fungal pathogens, including multidrug-resistant clinical isolates.
Collapse
Affiliation(s)
- Kiem Vu
- Department of Pharmacology, School of Medicine, University of California, Genome and Biomedical Sciences Facility, Davis, CA, United States
| | - Benjamin J. Buckley
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
| | - Richard S. Bujaroski
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- Monash Institute of Pharmaceutical Science (ATMCF), Monash University, Parkville, VIC, Australia
| | - Eduardo Blumwald
- Department of Plant Sciences, PRB Building, University of California, Davis, CA, Australia
| | - Michael J. Kelso
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
| | - Angie Gelli
- Department of Pharmacology, School of Medicine, University of California, Genome and Biomedical Sciences Facility, Davis, CA, United States
- *Correspondence: Angie Gelli,
| |
Collapse
|
22
|
Hoy MJ, Heitman J. Drug Target Elucidation Through Isolation and Analysis of Drug-Resistant Mutants in Cryptococcus neoformans. Methods Mol Biol 2023; 2658:127-143. [PMID: 37024699 PMCID: PMC10602406 DOI: 10.1007/978-1-0716-3155-3_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
Drug target identification is an essential component to antifungal drug development. Many methods, including large chemical library screening, natural product screening, and drug repurposing efforts, can identify compounds with favorable in vitro antifungal activity. However, these approaches will often identify compounds with no known mechanism of action. Herein, we describe a method utilizing the human fungal pathogen Cryptococcus neoformans to identify antifungal drug targets through the isolation of spontaneous resistant mutants, antifungal testing, whole-genome sequencing, and variant analysis.
Collapse
Affiliation(s)
- Michael J Hoy
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
| | - Joseph Heitman
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
23
|
The Trisubstituted Isoxazole MMV688766 Exerts Broad-Spectrum Activity against Drug-Resistant Fungal Pathogens through Inhibition of Lipid Homeostasis. mBio 2022; 13:e0273022. [PMID: 36300931 PMCID: PMC9765174 DOI: 10.1128/mbio.02730-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Candida species are among the most prevalent causes of systemic fungal infection, posing a growing threat to public health. While Candida albicans is the most common etiological agent of systemic candidiasis, the frequency of infections caused by non-albicans Candida species is rising. Among these is Candida auris, which has emerged as a particular concern. Since its initial discovery in 2009, it has been identified worldwide and exhibits resistance to all three principal antifungal classes. Here, we endeavored to identify compounds with novel bioactivity against C. auris from the Medicines for Malaria Venture's Pathogen Box library. Of the five hits identified, the trisubstituted isoxazole MMV688766 emerged as the only compound displaying potent fungicidal activity against C. auris, as well as other evolutionarily divergent fungal pathogens. Chemogenomic profiling, as well as subsequent metabolomic and phenotypic analyses, revealed that MMV688766 disrupts cellular lipid homeostasis, driving a decrease in levels of early sphingolipid intermediates and fatty acids and a concomitant increase in lysophospholipids. Experimental evolution to further probe MMV688766's mode of action in the model fungus Saccharomyces cerevisiae revealed that loss of function of the transcriptional regulator HAL9 confers resistance to MMV688766, in part through the upregulation of the lipid-binding chaperone HSP12, a response that appears to assist in tolerating MMV688766-induced stress. The novel mode of action we have uncovered for MMV688766 against drug-resistant fungal pathogens highlights the broad utility of targeting lipid homeostasis to disrupt fungal growth and how screening structurally-diverse chemical libraries can provide new insights into resistance-conferring stress responses of fungi. IMPORTANCE As widespread antimicrobial resistance threatens to propel the world into a postantibiotic era, there is a pressing need to identify mechanistically distinct antimicrobial agents. This is of particular concern when considering the limited arsenal of drugs available to treat fungal infections, coupled with the emergence of highly drug-resistant fungal pathogens, including Candida auris. In this work, we demonstrate that existing libraries of drug-like chemical matter can be rich resources for antifungal molecular scaffolds. We discovered that the small molecule MMV688766, from the Pathogen Box library, displays previously undescribed broad-spectrum fungicidal activity through perturbation of lipid homeostasis. Characterization of the mode of action of MMV688766 provided new insight into the protective mechanisms fungi use to cope with the disruption of lipid homeostasis. Our findings highlight that elucidating the genetic circuitry required to survive in the presence of cellular stress offers powerful insights into the biological pathways that govern this important phenotype.
Collapse
|
24
|
Fenley JDC, de Barros PP, do Carmo PHF, Garcia MT, Rossoni RD, Junqueira JC. Repurposing HIV Protease Inhibitors Atazanavir and Darunavir as Antifungal Treatments against Candida albicans Infections: An In Vitro and In Vivo Study. Curr Issues Mol Biol 2022; 44:5379-5389. [PMID: 36354676 PMCID: PMC9688711 DOI: 10.3390/cimb44110364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/21/2022] [Accepted: 10/27/2022] [Indexed: 09/01/2023] Open
Abstract
Candida albicans is the chief etiological agent of candidiasis, a mycosis prevalent in individuals with acquired immunodeficiency syndrome (AIDS). In recent years, the introduction of human immunodeficiency virus (HIV) protease inhibitors (HIV-PI) has reduced the prevalence of candidiasis in these patients. Seeking new therapeutic strategies based on the perspective of drug repositioning, we evaluated the effects of two second-generation HIV-PIs, atazanavir (ATV) and darunavir (DRV), on virulence factors of C. albicans and experimental candidiasis. For this, clinical strains of C. albicans were subjected to in vitro and in vivo treatments with ATV or DRV. As a result, ATV and DRV exhibited antifungal activity against fungal cells at 512 μg/mL, reduced the viability and biomass of biofilms, and inhibited filamentation of C. albicans. In addition, these HIV-PIs downregulated the expression of SAP2 and BRC1 genes of C. albicans. In an in vivo study, prophylactic use of ATV and DRV prolonged the survival rate of Galleria mellonella larvae infected with C. albicans. Therefore, ATV and DRV showed activity against C. albicans by reducing cell growth, biofilm formation, filamentation, and expression of virulence genes. Furthermore, ATV and DRV decreased experimental candidiasis, suggesting the repurposing of HIV-PIs as antifungal treatments for C. albicans infections.
Collapse
Affiliation(s)
- Juliana de C. Fenley
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (Unesp), São José dos Campos, São Paulo 12245-000, Brazil
| | - Patrícia P. de Barros
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (Unesp), São José dos Campos, São Paulo 12245-000, Brazil
- Multicampi School of Medical Sciences, Federal University of Rio Grande do Norte (UFRN), Caicó, Rio Grande do Norte 59300-000, Brazil
| | - Paulo H. F. do Carmo
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (Unesp), São José dos Campos, São Paulo 12245-000, Brazil
| | - Maíra T. Garcia
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (Unesp), São José dos Campos, São Paulo 12245-000, Brazil
| | - Rodnei D. Rossoni
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (Unesp), São José dos Campos, São Paulo 12245-000, Brazil
| | - Juliana C. Junqueira
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (Unesp), São José dos Campos, São Paulo 12245-000, Brazil
| |
Collapse
|
25
|
de Siqueira VM, da Silva BGM, Passos JCDS, Pinto AP, da Rocha JBT, Alberto-Silva C, Costa MS. (MeOPhSe)2, a synthetic organic selenium compound, inhibits virulence factors of Candida krusei: Adherence to cervical epithelial cells and biofilm formation. J Trace Elem Med Biol 2022; 73:127019. [PMID: 35709560 DOI: 10.1016/j.jtemb.2022.127019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 02/26/2022] [Accepted: 06/07/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Systemic candidiasis is produced by Candida albicans or non-albicans Candida species, opportunistic fungi that produce both superficial and invasive infections. Despite the availability of a wide range of antifungal agents for the treatment of candidiasis, failure of therapy is observed frequently, which opens new avenues in the field of alternative therapeutic strategies. METHODS The effects of p,p'-methoxyl-diphenyl diselenide [(MeOPhSe)2], a synthetic organic selenium (organochalcogen) compound, were investigated on virulence factors of C. krusei and compared with its antifungal effects on the virulence factors related to adhesion to cervical epithelial cell surfaces with C. albicans. RESULTS (MeOPhSe)2, a compound non-toxic in epithelial (HeLa) and fibroblastic (Vero) cells, inhibited the growth in a dose-dependent manner and changed the kinetics parameters of C. krusei and, most importantly, extending the duration of lag phase of growth, inhibiting biofilm formation, and changing the structure of biofilm. Also, (MeOPhSe)2 reduced C. albicans and C. krusei adherence to cervical epithelial cells, an important factor for the early stage of the Candida-host interaction. The reduction was 37.24 ± 2.7 % in C. krusei (p = 0.00153) and 32.84 ± 3.2 % in C. albicans (p = 0.0072) at 20 µM (MeOPhSe)2, and the effect is in a concentration-dependent manner. Surprisingly, the antifungal potential on adhesion was similar between both species, indicating the potential of (MeOPhSe)2 as a promising antifungal drug against different Candida infections. CONCLUSION Overall, we demonstrated the potential of (MeOPhSe)2 as an effective antifungal drug against the virulence factors of Candida species.
Collapse
Affiliation(s)
- Victor Mendes de Siqueira
- Instituto de Pesquisa e Desenvolvimento - IP&D, Universidade do Vale do Paraíba - UNIVAP, Av. Shishima Hifumi, São José dos Campos, SP 2911, Brazil
| | - Bruna Graziele Marques da Silva
- Instituto de Pesquisa e Desenvolvimento - IP&D, Universidade do Vale do Paraíba - UNIVAP, Av. Shishima Hifumi, São José dos Campos, SP 2911, Brazil
| | - Juliene Cristina da Silva Passos
- Instituto de Pesquisa e Desenvolvimento - IP&D, Universidade do Vale do Paraíba - UNIVAP, Av. Shishima Hifumi, São José dos Campos, SP 2911, Brazil
| | - Ana Paula Pinto
- Instituto de Pesquisa e Desenvolvimento - IP&D, Universidade do Vale do Paraíba - UNIVAP, Av. Shishima Hifumi, São José dos Campos, SP 2911, Brazil
| | | | - Carlos Alberto-Silva
- Natural and Humanities Sciences Center, Experimental Morphophysiology Laboratory Federal University of ABC (UFABC), Rua Arcturus, no 03, Bloco Delta, São Bernardo do Campo, SP 09606-070, Brazil
| | - Maricilia Silva Costa
- Instituto de Pesquisa e Desenvolvimento - IP&D, Universidade do Vale do Paraíba - UNIVAP, Av. Shishima Hifumi, São José dos Campos, SP 2911, Brazil.
| |
Collapse
|
26
|
Abduljalil H, Bakri A, Albashaireh K, Alshanta OA, Brown JL, Sherry L, Kean R, Nile C, McLean W, Ramage G. Screening the Tocriscreen™ bioactive compound library in search for inhibitors of
Candida
biofilm formation. APMIS 2022; 130:568-577. [PMID: 35791082 PMCID: PMC9541805 DOI: 10.1111/apm.13260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/29/2022] [Indexed: 11/28/2022]
Abstract
Biofilms formed by Candida species present a significant clinical problem due to the ineffectiveness of many conventional antifungal agents, in particular the azole class. We urgently require new and clinically approved antifungal agents quickly for treatment of critically ill patients. To improve efficiency in antifungal drug development, we utilized a library of 1280 biologically active molecules within the Tocriscreen 2.0 Micro library. Candida aurisNCPF 8973 and Candida albicansSC5314 were initially screened for biofilm inhibitory activity using metabolic and biomass quantitative assessment methods, followed up by targeted evaluation of five selected hits. The initial screening (80% metabolic inhibition rate) revealed that there was 90 and 87 hits (approx. 7%) for C. albicans and C. auris, respectively. Additionally, all five compounds selected from the initial hits exhibited a biofilm inhibition effect against several key Candida species tested, including C. glabrata and C. krusei. Toyocamycin displayed the most potent activity at concentrations as low as 0.5 μg/mL, though was limited to inhibition. Darapladib demonstrated an efficacy for biofilm inhibition and treatment at a concentration range from 8 to 32 μg/mL and from 16 to 256 μg/mL, respectively. Combinational testing with conventional antifungals against C. albicans strains demonstrated a range of synergies for planktonic cells, and notably an anti‐biofilm synergy for darapladib and caspofungin. Together, these data provide new insights into antifungal management possibilities for Candida biofilms.
Collapse
Affiliation(s)
- Hafsa Abduljalil
- Glasgow Biofilm Research Group School of Medicine Dentistry and Nursing College of Medical, Veterinary and Life Sciences University of Glasgow Glasgow UK
| | - Ahmed Bakri
- Glasgow Biofilm Research Group School of Medicine Dentistry and Nursing College of Medical, Veterinary and Life Sciences University of Glasgow Glasgow UK
| | - Khawlah Albashaireh
- Glasgow Biofilm Research Group School of Medicine Dentistry and Nursing College of Medical, Veterinary and Life Sciences University of Glasgow Glasgow UK
| | - Om Alkhir Alshanta
- Glasgow Biofilm Research Group School of Medicine Dentistry and Nursing College of Medical, Veterinary and Life Sciences University of Glasgow Glasgow UK
| | - Jason L. Brown
- Glasgow Biofilm Research Group School of Medicine Dentistry and Nursing College of Medical, Veterinary and Life Sciences University of Glasgow Glasgow UK
| | - Leighann Sherry
- Glasgow Biofilm Research Group School of Medicine Dentistry and Nursing College of Medical, Veterinary and Life Sciences University of Glasgow Glasgow UK
| | - Ryan Kean
- Department of Biological and Biomedical Sciences Glasgow Caledonian University
| | | | - William McLean
- Glasgow Biofilm Research Group School of Medicine Dentistry and Nursing College of Medical, Veterinary and Life Sciences University of Glasgow Glasgow UK
| | - Gordon Ramage
- Glasgow Biofilm Research Group School of Medicine Dentistry and Nursing College of Medical, Veterinary and Life Sciences University of Glasgow Glasgow UK
| |
Collapse
|
27
|
Donlin MJ, Meyers MJ. Repurposing and optimization of drugs for discovery of novel antifungals. Drug Discov Today 2022; 27:2008-2014. [PMID: 35489676 PMCID: PMC11182377 DOI: 10.1016/j.drudis.2022.04.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 12/17/2022]
Abstract
Although fungal diseases are a major and growing public health concern, there are only four major classes of drug to treat primary fungal pathogens. The pipeline of new antifungals in clinical development is relatively thin compared with other disease classes. One approach to rapidly identify and provide novel treatment options is to repurpose existing drugs as antifungals. However, such proposed drug-repurposing candidates often suffer suboptimal efficacy and pharmacokinetics (PK) for fungal diseases. Herein, we briefly review the current antifungal drug pipeline and recent approaches to optimize existing drugs into novel molecules with unique modes of action relative to existing antifungal drug classes.
Collapse
Affiliation(s)
- Maureen J Donlin
- Edward. A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St Louis, MO, USA; Saint Louis University Institute for Drug and Biotherapeutic Innovation, USA.
| | - Marvin J Meyers
- Department of Chemistry, Saint Louis University, St Louis, MO, USA; Saint Louis University Institute for Drug and Biotherapeutic Innovation, USA
| |
Collapse
|
28
|
Breuer MR, Dasgupta A, Vasselli JG, Lin X, Shaw BD, Sachs MS. The Antidepressant Sertraline Induces the Formation of Supersized Lipid Droplets in the Human Pathogen Cryptococcus neoformans. J Fungi (Basel) 2022; 8:jof8060642. [PMID: 35736125 PMCID: PMC9224953 DOI: 10.3390/jof8060642] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 01/11/2023] Open
Abstract
The prevalence and increasing incidence of fungal infections globally is a significant worldwide health problem. Cryptococcosis, primarily caused by the pathogenic yeast Cryptococcus neoformans, is responsible for approximately 181,000 estimated deaths annually. The scarcity of treatments and the increasing resistance to current therapeutics highlight the need for the development of antifungal agents which have novel mechanisms of action and are suitable for clinical use. Repurposing existing FDA-approved compounds as antimycotic therapeutics is a promising strategy for the rapid development of such new treatments. Sertraline (SRT), a commonly prescribed antidepressant, is a broad-spectrum antifungal agent with particular efficacy against C. neoformans. However, the effect of SRT on fungal physiology is not understood. Here, we report that SRT induces the formation of supersized lipid droplets (SLDs) in C. neoformans, and in Candida albicans, Saccharomyces cerevisiae, and Aspergillus fumigatus. SLDs were not induced in C. neoformans by treatment with the antifungal fluconazole (FLC), consistent with SRT and FLC acting differently to perturb C. neoformans physiology. The formation of SLDs in response to SRT indicates that this compound alters the lipid metabolism of C. neoformans. Moreover, the SRT-induced enlargement of LDs in other fungal species may indicate a common fungal response to SRT.
Collapse
Affiliation(s)
- Matthew R. Breuer
- Department of Biology, Texas A&M University, College Station, TX 77843, USA; (M.R.B.); (A.D.)
| | - Ananya Dasgupta
- Department of Biology, Texas A&M University, College Station, TX 77843, USA; (M.R.B.); (A.D.)
| | - Joseph G. Vasselli
- Department of Plant Pathology and Microbiology, Texas A&M University, College Station, TX 77843, USA; (J.G.V.); (B.D.S.)
| | - Xiaorong Lin
- Department of Microbiology, University of Georgia, Athens, GA 30602, USA;
| | - Brian D. Shaw
- Department of Plant Pathology and Microbiology, Texas A&M University, College Station, TX 77843, USA; (J.G.V.); (B.D.S.)
| | - Matthew S. Sachs
- Department of Biology, Texas A&M University, College Station, TX 77843, USA; (M.R.B.); (A.D.)
- Correspondence: ; Tel.: +1-(979)-845-5930
| |
Collapse
|
29
|
da Silva BGM, Pinto AP, Passos JCDS, da Rocha JBT, Alberto-Silva C, Costa MS. Diphenyl diselenide suppresses key virulence factors of Candida krusei, a neglected fungal pathogen. BIOFOULING 2022; 38:427-440. [PMID: 35670068 DOI: 10.1080/08927014.2022.2084388] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 05/17/2022] [Accepted: 05/25/2022] [Indexed: 06/15/2023]
Abstract
Candida krusei is a candidiasis etiological agent of relevance in the clinical setting because of its intrinsic resistance to fluconazole. Also, it has opened up new paths in the area of alternative therapeutic techniques. This project demonstrated the effects of diphenyl diselenide (PhSe)2 and p-cloro diphenyl diselenide (pCl-PhSe)2, two organochalcogen compounds, on relevant virulence factors for the early stage of the C. krusei host interaction and infection process. Both compounds inhibited adherence of C. krusei to both polystyrene surfaces and cervical epithelial cells and biofilm formation; the structure of the biofilm was also changed in a dose-dependent manner. In addition, both compounds inhibited C. krusei growth, but (PhSe)2 significantly increased the time duration of the lag phase and delayed the start of the exponential phase in growth kinetics. (PhSe)2 has more potential antifungal activity than (pCl-PhSe)2 in inhibiting the adherence to epithelial cells, biofilm formation, and growth of C. krusei.
Collapse
Affiliation(s)
| | - Ana Paula Pinto
- Instituto de Pesquisa e Desenvolvimento-IP&D, Universidade do Vale do Paraíba, São José dos Campos, São Paulo, Brazil
| | | | - João Batista Teixeira da Rocha
- Departamento de Química, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, São Paulo, Brazil
| | - Carlos Alberto-Silva
- Experimental Morphophysiology Laboratory, Natural and Humanities Sciences Center (CCNH), Federal University of ABC-UFABC, São Paulo, Brazil
| | - Maricilia Silva Costa
- Instituto de Pesquisa e Desenvolvimento-IP&D, Universidade do Vale do Paraíba, São José dos Campos, São Paulo, Brazil
| |
Collapse
|
30
|
Li W, Yun Z, Ji C, Tu J, Yang W, Li J, Liu N, Sheng C. Discovery of Novel Sertraline Derivatives as Potent Anti- Cryptococcus Agents. J Med Chem 2022; 65:6541-6554. [DOI: 10.1021/acs.jmedchem.1c01845] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Wang Li
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Zhaolin Yun
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Changjin Ji
- School of Pharmacy, East China University of Science & Technology, 130 Meilong Road, Shanghai 200237, People’s Republic of China
| | - Jie Tu
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Wanzhen Yang
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Jian Li
- School of Pharmacy, East China University of Science & Technology, 130 Meilong Road, Shanghai 200237, People’s Republic of China
| | - Na Liu
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Chunquan Sheng
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| |
Collapse
|
31
|
In vitro study on the potential fungicidal effects of atorvastatin in combination with some azole drugs against multidrug resistant Candida albicans. World J Microbiol Biotechnol 2021; 37:191. [PMID: 34632522 PMCID: PMC8502632 DOI: 10.1007/s11274-021-03158-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 09/30/2021] [Indexed: 01/08/2023]
Abstract
The resistance of Candida albicans to azole drugs represents a great global challenge. This study investigates the potential fungicidal effects of atorvastatin (ATO) combinations with fluconazole (FLU), itraconazole (ITR), ketoconazole (KET) and voriconazole (VOR) against thirty-four multidrug-resistant (MDR) C. albicans using checkerboard and time-kill methods. Results showed that 94.12% of these isolates were MDR to ≥ two azole drugs, whereas 5.88% of them were susceptible to azole drugs. The tested isolates exhibited high resistance rates to FLU (58.82%), ITR (52.94%), VOR (47.06%) and KET (35.29%), whereas only three representative (8.82%) isolates were resistant to all tested azoles. Remarkably, the inhibition zones of these isolates were increased at least twofold with the presence of ATO, which interacted in a synergistic (FIC index ≤ 0.5) manner with tested azoles. In silico docking study of ATO and the four azole drugs were performed against the Lanosterol 14-alpha demethylase enzyme (ERG11) of C. albicans. Results showed that the mechanism of action of ATO against C. albicans is similar to that of azole compounds, with a docking score (−4.901) lower than azole drugs (≥5.0) due to the formation a single H-bond with Asp 225 and a pi–pi interaction with Thr 229. Importantly, ATO combinations with ITR, VOR and KET achieved fungicidal effects (≥ 3 Log10 cfu/ml reduction) against the representative isolates, whereas a fungistatic effect (≤ 3 Log10 cfu/ml reduction) was observed with FLU combination. Thus, the combination of ATO with azole drugs could be promising options for treating C. albicans infection.
Collapse
|
32
|
|
33
|
Pyrogallol and Fluconazole Interact Synergistically In Vitro against Candida glabrata through an Efflux-Associated Mechanism. Antimicrob Agents Chemother 2021; 65:e0010021. [PMID: 33875436 PMCID: PMC8373228 DOI: 10.1128/aac.00100-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Candida glabrata is currently the first or second most commonly encountered non-albicans Candida species worldwide. The potential severity of Candida resistance mandates the discovery of novel antifungal agents, including those that can be used in combination therapies. In this study, we evaluated the in vitro interactions of pyrogallol (PG) and azole drugs against 22 clinical C. glabrata isolates. The potential mechanism underlying the synergism between PG and fluconazole (FLC) was investigated by the rhodamine 6G efflux method and quantitative reverse transcription (qRT)-PCR analysis. In susceptibility tests, PG showed strong synergism with FLC, itraconazole (ITC), and voriconazole (VRC), with fractional inhibitory concentration index values of 0.18 to 0.375 for PG+FLC, 0.250 to 0.750 for PG+ITC, and 0.141 to 0.750 for PG+VRC. Cells grown in the presence of PG+FLC exhibited reduced rhodamine 6G extrusion and significantly downregulated expression of the efflux-related genes CgCDR1, CgCDR2, and CgPDR1 compared with cells grown in the presence of PG or FLC alone. PG did not potentiate FLC when tested against a ΔCgpdr1 strain. Restoration of a functional CgPDR1 allele also restored the synergism. These results indicate that PG is an antifungal agent that synergistically potentiates the activity of azoles. Furthermore, PG appears to exert its effects by inhibiting efflux pumps and downregulating CgCDR1, CgCDR2, and CgPDR1, with CgPDR1 probably playing a crucial role in this process.
Collapse
|
34
|
Wall G, Chen E, Hull MV, Lopez-Ribot JL. Screening the CALIBR ReFRAME Library in Search for Inhibitors of Candida auris Biofilm Formation. Front Cell Infect Microbiol 2020; 10:597931. [PMID: 33324579 PMCID: PMC7723901 DOI: 10.3389/fcimb.2020.597931] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 11/02/2020] [Indexed: 12/14/2022] Open
Abstract
Candida auris is an emerging yeast which, since its first isolation about a decade ago, has spread rapidly and triggered major infectious outbreaks in health care facilities around the world. C. auris strains often display resistance to clinically-used antifungal agents, contributing to high mortality rates. Thus, there is an urgent need for new antifungals to contain the spread of this emerging multi-drug resistant pathogen and to improve patient outcomes. However, the timeline for the development of a new antifungal agent typically exceeds 10‑15 years. Thus, repurposing of current drugs could significantly accelerate the development and eventual deployment of novel therapies for the treatment of C. auris infections. Toward this end, in this study we have profiled a library of known drugs encompassing approximately 12,000 clinical-stage or FDA-approved small molecules in search for known molecules with antifungal activity against C. auris; more specifically, those capable of inhibiting C. auris biofilm formation. From this library, 100 compounds displaying antifungal activity were identified in the initial screen, including 26 compounds for which a dose-response relationship with biofilm-inhibitory activity against C. auris could be confirmed. Of these, five were identified as the most interesting potential repositionable candidates. Due to their known pharmacological and human safety profiles, identification of such compounds should allow for their accelerated preclinical and clinical development for the treatment of C. auris infections.
Collapse
Affiliation(s)
- Gina Wall
- Department of Biology and The South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, United States
| | - Emily Chen
- Calibr, a division of The Scripps Research Institute, La Jolla, CA, United States
| | - Mitchell V Hull
- Calibr, a division of The Scripps Research Institute, La Jolla, CA, United States
| | - Jose L Lopez-Ribot
- Department of Biology and The South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, United States
| |
Collapse
|
35
|
Current Antimycotics, New Prospects, and Future Approaches to Antifungal Therapy. Antibiotics (Basel) 2020; 9:antibiotics9080445. [PMID: 32722455 PMCID: PMC7460292 DOI: 10.3390/antibiotics9080445] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 12/11/2022] Open
Abstract
Fungal infections represent an increasing threat to a growing number of immune- and medically compromised patients. Fungi are eukaryotic organisms and, as such, there is a limited number of selective targets that can be exploited for antifungal drug development. This has also resulted in a very restricted number of antifungal drugs that are clinically available for the treatment of invasive fungal infections at the present time—polyenes, azoles, echinocandins, and flucytosine. Moreover, the utility of available antifungals is limited by toxicity, drug interactions and the emergence of resistance, which contribute to high morbidity and mortality rates. This review will present a brief summary on the landscape of current antifungals and those at different stages of clinical development. We will also briefly touch upon potential new targets and opportunities for novel antifungal strategies to combat the threat of fungal infections.
Collapse
|